EP4356933A1 - Zusammensetzung zur in-vivo-freisetzung von mrna mit modifiziertem nukleotid - Google Patents

Zusammensetzung zur in-vivo-freisetzung von mrna mit modifiziertem nukleotid Download PDF

Info

Publication number
EP4356933A1
EP4356933A1 EP23814344.0A EP23814344A EP4356933A1 EP 4356933 A1 EP4356933 A1 EP 4356933A1 EP 23814344 A EP23814344 A EP 23814344A EP 4356933 A1 EP4356933 A1 EP 4356933A1
Authority
EP
European Patent Office
Prior art keywords
glycero
mrna
composition according
composition
propane
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP23814344.0A
Other languages
English (en)
French (fr)
Inventor
Yang Je Cho
Seok Hyun Kim
Kwangsung KIM
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eyegene Inc
Original Assignee
Eyegene Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020230102612A external-priority patent/KR20240035322A/ko
Application filed by Eyegene Inc filed Critical Eyegene Inc
Publication of EP4356933A1 publication Critical patent/EP4356933A1/de
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars

Definitions

  • the present invention relates to a composition for in-vivo delivering mRNA containing a modified nucleotide, and more particularly to a composition for in-vivo delivering mRNA containing a modified nucleotide, which includes liposomes based on a cationic lipid.
  • Gene therapy and genetic vaccination are technologies that have already been proven in the field of medicine and are generally applied, and may be used to treat not only genetic diseases, but also autoimmune diseases, infectious diseases, cancer or tumorrelated diseases, inflammatory diseases, and the like.
  • DNA and RNA may be used as nucleic acid molecules for gene administration, and it is known that DNA is relatively stable and easy to handle compared to RNA.
  • DNA a potential risk may arise if the DNA segment administered into the patient's genome is inserted at an undesirable location and the gene is damaged. Additionally, unwanted anti-DNA antibodies may appear, and another problem is that the level of the peptide or protein expressed by transcription/translation after DNA administration is limited.
  • the presence or absence of a specific transcription factor that regulates DNA transcription has a major impact on the expression level of the administered DNA, and in the absence of a specific transcription factor, a sufficient amount of RNA is not produced by DNA transcription, and consequently, the level of peptide or protein produced by translation is also limited.
  • RNA when RNA is used as a tool for gene administration, RNA does not require transcription and is thus able to directly synthesize proteins in the cytoplasm without the need to enter the nucleus like DNA, so there is no fear of intruding into the cell chromosome and causing unwanted genetic damage. Moreover, RNA has a shorter half-life than DNA and thus does not induce longterm genetic modification ( Sayour EJ, et al., J. Immunother. Cancer 2015; 3:13, 2015 ). When delivered into cells, a general RNA vaccine is activated for a short period of time to express a target protein and is destroyed by an enzymatic reaction within a few days, and a specific immune response to the expressed target antigen (protein) remains.
  • RNA when using RNA as a tool for gene administration, RNA works only when it passes through the cell membrane without the need to pass through the nuclear membrane, making it possible to express the target protein in a similar level as when using DNA even in a smaller amount than DNA. Moreover, RNA itself has immunereinforcing activity and is thus capable of exhibiting the same immune effect even when administered in a small amount compared to DNA.
  • RNA is a highly unstable molecular species that may be readily degraded by ubiquitous RNases.
  • nucleic acid delivery to obtain a desired response in a biological system.
  • Nucleic acid-based therapeutics hold tremendous promise, but realizing this promise requires effective delivery of nucleic acids to appropriate regions within cells or organisms.
  • nucleic acids for therapeutic and prophylactic purposes currently faces two problems.
  • Incorporating lipid nanoparticles formed from cationic lipids and other lipid components such as neutral lipids, cholesterol, PEG, pegylated lipids, and oligonucleotides has been attempted to block degradation of RNA in blood and promote cellular uptake of nucleic acids.
  • mRNA When a delivery carrier based on lipids such as liposomes or lipid nanoparticles is used, mRNA is usually adsorbed outside or encapsulated inside. In particular, when mRNA is adsorbed to the outside of the delivery carrier, it is known to use an aggregate of liposomes and a nucleic acid, rather than liposomes alone. Since the combination of lipids, the state of the nucleic acid, and the ratio of nucleic acid to liposomes may affect adsorption capacity and stability, optimization therefor is required.
  • a modified nucleotide has high expression efficiency in vivo compared to mRNA using an unmodified nucleotide ( US8278036B2 , KR10-2171849B1 , KR10-2014601B1 ).
  • the modified nucleotide may include pseudouridine, N1-methyl-pseudouridine, etc., but it is necessary to optimize the combination of the delivery carrier and the modified nucleotide.
  • the present inventors have made great efforts to find techniques for improving a delivery carrier capable of inducing stable protein expression by stably delivering mRNA containing a modified nucleotide in vivo to increase intracellular expression efficiency, and thus ascertained that [liposome + mRNA complex] based on cationic liposomes and mRNA containing a specific modified nucleotide may exhibit low inflammatory response and high expression efficiency in vivo, thereby culminating in the present invention.
  • It is yet another object of the present invention to provide a functional cosmetic composition including the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient.
  • the present invention provides a composition for delivering mRNA containing a modified nucleotide, including liposomes based on a cationic lipid.
  • the present invention provides a pharmaceutical composition for preventing or treating a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection, including the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient.
  • the present invention provides a vaccine including the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient.
  • the present invention provides a functional cosmetic composition including the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient.
  • the present invention provides a method of preventing or treating a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection, including administering a composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid.
  • the present invention provides the use of a composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid for preventing or treating a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection.
  • a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection.
  • the present invention provides the use of a composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid for the manufacture of a medicament for preventing or treating a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection.
  • a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection.
  • the present invention is intended to provide a method that exhibits a stable effect by increasing intracellular expression efficiency of mRNA. Therefore, a composition for delivering mRNA using liposomes based on a cationic lipid is prepared, confirming low inflammatory response and high expression efficiency in vivo.
  • composition for delivering mRNA containing a specific type of modified nucleotide is confirmed to be vastly superior.
  • mRNA may be provided in the form of a complex with liposomes based on a cationic lipid, and accordingly, the composition for delivering mRNA using liposomes based on a cationic lipid is interchangeably used with [liposome + mRNA complex] .
  • An aspect of the present invention pertains to a composition for delivering mRNA containing a modified nucleotide, which includes liposomes based on a cationic lipid.
  • the modified nucleotide may include at least one backbone-modified, sugar-modified, or base-modified nucleotide, but is not limited thereto.
  • a modified nucleoside or nucleotide that may be incorporated into a modified mRNA compound including mRNA sequences as described herein may be modified at the sugar moiety.
  • a 2' hydroxyl group (OH) may be modified or replaced with a number of different "oxy" or “deoxy” substituents.
  • R H, alkyl, cycloalkyl, aryl, aralkyl, hetero
  • the "deoxy" modification may include hydrogen or amino (e.g. NH 2 ; alkylamino, dialkylamino, heterocyclyl, arylamino, diarylamino, heteroarylamino, diheteroarylamino, or amino acid); or an amino group may be attached to the sugar through a linker, in which the linker includes at least one selected from among atoms C, N, and O.
  • modified mRNA may include a nucleotide containing arabinose, such as sugar.
  • a modified nucleoside or nucleotide that may be incorporated into a modified mRNA compound including mRNA sequences as described herein, the phosphate backbone may be further modified.
  • the phosphate group of the backbone may be modified by replacing one or more oxygen atoms with other substituents.
  • a modified nucleoside or nucleotide may include the complete replacement of an unmodified phosphate moiety with a modified phosphate described herein.
  • modified phosphate group may include, but are not limited to, phosphorothioate, phosphoroselenate, boranophosphate, boranophosphate ester, hydrogen phosphonate, phosphoroamidate, alkyl or aryl phosphonate, and phosphotriester.
  • Phosphorodithioate has both unlinked oxygens replaced by sulfur.
  • a phosphate linker may also be modified by substitution of linked oxygen with nitrogen (crosslinked phosphoroamidate), sulfur (crosslinked phosphorothioate), or carbon (crosslinked methylenephosphonate).
  • a modified nucleoside or nucleotide that may be incorporated into a modified mRNA compound including mRNA sequences as described herein may be further modified at the nucleobase moiety.
  • the nucleobase found in mRNA include, but are not limited to, adenine, guanine, cytosine, and uracil.
  • the nucleoside or nucleotide described herein may be chemically modified at the major groove face.
  • major groove chemical modifications may include amino groups, thiol groups, alkyl groups, or halo groups.
  • the nucleotide analogue/modification is selected from among base modifications such as 2-amino-6-chloropurine riboside-5'-triphosphate, 2-aminopurine-riboside-5'-triphosphate, 2-aminoadenosine-5'-triphosphate, 2'-amino-2'-deoxycytidine-triphosphate, 2-thiocytidine-5'-triphosphate, 2-thiouridine-5'-triphosphate, 2'-fluorothymidine-5'-triphosphate, 2'-O-methyl-inosine-5'-triphosphate, 4-thiouridine-5'-triphosphate, 5-aminoallylcytidine-5'-triphosphate, 5-aminoallyluridine-5'-triphosphate, 5-bromocytidine-5'-triphosphate, 5-bromouridine-5'-triphosphate, 5-bromo-2'-deoxycytidine-5'-triphosphate, 5-
  • the modified nucleoside may include pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyl-uridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, 1-taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-1-methyl-pseudouridine, 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-
  • the modified nucleoside may include 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-1-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza-pseudoisocytidine, 1-methyl-1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2-thio-zebul
  • the modified nucleoside may include 2-aminopurine, 2,6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyladenosine, N6-methyladenosine, N6-isopentyladenosine, N6-(cis-hydroxyisopentyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl)adenosine, N6-glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-threonylcarbamoyladenosine, N6,N6-d
  • the modified nucleoside may include inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, 1-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, or N2,N2-dimethyl-6-thio-guanosine.
  • the nucleotide may be modified at the major groove face and may include replacement of hydrogen with a methyl group or halo group at C-5 of uracil.
  • the modified nucleoside is 5'-O-(1-thiophosphate)-adenosine, 5'-O-(1-thiophosphate)-cytidine, 5'-O-(1-thiophosphate)-guanosine, 5'-O-(1-thiophosphate)-uridine, or 5'-O-(1-thiophosphate)-pseudouridine.
  • the modified mRNA may include any nucleoside modification selected from among 6-aza-cytidine, 2-thio-cytidine, ⁇ -thio-cytidine, pseudoisocytidine, 5-aminoallyl-uridine, 5-iodo-uridine, N1-methyl-pseudouridine, 5,6-dihydrouridine, ⁇ -thio-uridine, 4-thio-uridine, 6-aza-uridine, 5-hydroxy-uridine, deoxy-thymidine, 5-methyl-uridine, pyrrolo-cytidine, inosine, ⁇ -thio-guanosine, 6-methyl-guanosine, 5-methyl-cytidine, 8-oxo-guanosine, 7-deaza-guanosine, N1-methyl-adenosine, 2-amino-6-chloro-purine, N6-methyl-2-amino-purine, 6-chloro-purine, N6-methyl-a
  • the modified nucleotide is selected from the group consisting of pseudouridine ( ⁇ ), N1-methylpseudouridine (m1 ⁇ ), 5-methyluridine (m5U), 2-thiouridine (s2U), 2'-O-methyl-uridine (Um), 5-methylcytidine (m5C), and 5-methoxyuridine (5moU), and is most preferably 5-methoxyuridine (5moU), but is not limited thereto.
  • the cationic lipid includes a lipid that is continuously cationic without the influence of pH change or an ionic lipid that is converted to be cationic by pH change.
  • the cationic lipid is preferably at least one selected from the group consisting of dimethyldioctadecylammonium bromide (DDA), C12-200, 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), 3 ⁇ -[N-(N',N'-dimethylaminoethane)carbamoyl cholesterol (DC-Chol), 1,2-dioleoyloxy-3-dimethylammonium propane (DODAP), 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA), 1,2-dimyristoleoyl-sn-glycero-3-ethylphosphocholine (14:1 Ethyl PC), 1-palmitoyl-2-oleoyl-sn-glycero-3-ethylphosphocholine (16:0/18:1 Ethyl PC), 1,2-dioleoyl-sn-glycerammonium propane
  • DOTAP dioleoyl-3-trimethylammonium propane
  • DOTAP dioleoyl-3-trimethylammonium propane
  • DOTAP dioleoyl-3-trimethylammonium propane
  • the composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid may further include a neutral lipid.
  • the neutral lipid includes a lipid that is continuously neutral without the influence of pH change or an ionic lipid that is converted to be neutral by pH change.
  • the neutral lipid may be selected from the group consisting of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), phosphatidylserine (PS), phosphoethanolamine (PE), phosphatidylglycerol (PG), phosphoric acid (PA), phosphatidylcholine (PC), 1,2-dioleoyl-sn-glycero-3-phospho-(1'-myo-inositol
  • DOPE
  • DOPE 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine
  • a preferred neutral lipid has the structure of Chemical Formula 2 below and is used as an auxiliary lipid for forming cationic liposomes or lipid nanoparticles.
  • the liposomes according to the present invention may further include at least one delivery factor selected from the group consisting of protamine, albumin, transferrin, PTD (protein transduction domain), CPP (cell-penetrating peptide), PEG (polyethylene glycol), pegylated lipid, metal ion-linked lipid, and macrophage targeting moiety.
  • at least one delivery factor selected from the group consisting of protamine, albumin, transferrin, PTD (protein transduction domain), CPP (cell-penetrating peptide), PEG (polyethylene glycol), pegylated lipid, metal ion-linked lipid, and macrophage targeting moiety.
  • the weight ratio of cationic lipid to neutral lipid is 1:9 to 9.5:0.5, preferably 2:8 to 9:1, more preferably 3:7 to 8:2, most preferably 4:6 to 7:3, but is not limited thereto.
  • composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid may further include cholesterol.
  • the weight ratio of cationic lipid to cholesterol is 6:1 to 1:3, preferably 4:1 to 1.0:2.5, more preferably 3:1 to 1:2, most preferably 2.5:1.0 to 1.0:1.5, but is not limited thereto.
  • the weight ratio of cationic lipid to neutral lipid to cholesterol is 1.0-9.5:0.5-9.0:0.05-3.00, preferably 3-8:7-1:0.45-7.00, more preferably 1.0-3.5:1.0-3.5:0.5-3.0, but is not limited thereto.
  • the weight ratio of cationic lipid to neutral lipid to cholesterol is set to 2:2:1 (40:40:20, w/w/w), but is not limited thereto.
  • cholesterol may be mixed in a weight ratio of 0.20-0.85, preferably 0.4-0.6 relative to DOTAP to form liposomes.
  • the mixing ratio of liposomes and mRNA may be represented as N:P ratio, and the N:P ratio affects mRNA expression and stability of the composition.
  • the N:P ratio of liposomes or lipid nanoparticles and mRNA may be 0.2:1.0 to 1.4:1.0, preferably 0.23:1.00 to 1.0:1.0, more preferably 0.46:1.00 to 1.0:1.0, but is not limited thereto. In an exemplary embodiment of the present invention, the N:P ratio at 0.6:1.0 is used.
  • the composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid may further include an immune enhancer, but the use of an immune enhancer is not essential, and a sufficient vaccine effect is exhibited even in the absence of an immune enhancer.
  • the immune enhancer usable in the present invention is an immune enhancer selected from the group consisting of a material responding to a pattern recognition receptor (PRR) corresponding to a pathogen-associated molecular pattern (PAMP), CpG DNA, lipoprotein, flagella, poly I:C, saponin, squalene, tricaprin, 3D-MPL, and detoxified lipooligosaccharide (dLOS), but is not limited thereto.
  • the detoxified lipooligosaccharide may be a material disclosed in Korean Patent No. 1509456 or Korean Patent No. 2042993 , but the present invention is not limited thereto.
  • mRNA is capable of encoding a peptide or protein that may act as an immunogen.
  • mRNA may be mRNA having at least one open reading frame (ORF) that may be translated by a cell or organism, which is typically provided along with mRNA.
  • the product of this translation is an antigen, preferably a peptide or protein acting as an immunogen.
  • Such a product may also be a fusion protein composed of two or more immunogens, for example, a fusion protein composed of two or more epitopes, peptides, or proteins derived from the same or different viral proteins, in which the epitopes, peptides, or proteins may be connected by a linker sequence.
  • mRNA may be understood to be synthetic mRNA, namely an mRNA molecule that does not occur naturally.
  • a synthetic mRNA molecule may be understood as a non-natural mRNA molecule.
  • Such an mRNA molecule may be non-natural due to (non-naturally occurring) individual sequences and/or other non-naturally occurring alterations, such as structural alterations of nucleotides.
  • a synthetic mRNA molecule may be designed and/or produced by genetic engineering methods corresponding to a desired synthetic sequence (heterologous sequence) of nucleotide.
  • mRNA may exhibit modifications that increase resistance to degradation in vivo (e.g. degradation by exo- or endo-nucleases) and/or degradation in vitro (e.g. by the preparation process before administration of a vaccine, for example, in the process of preparing a vaccine solution to be administered).
  • Stabilization of RNA may be achieved, for example, by providing a 5'-CAP structure, poly-A-tail, or any other UTR modification.
  • Stabilization of RNA may also be achieved by chemical modification or modification of G/C content of nucleic acids. A variety of other methods are known in the art and are applicable to the present invention.
  • composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid may be in the form in which mRNA containing a modified nucleotide is encapsulated inside liposomes or in which mRNA containing a modified nucleotide additionally binds to the outside as well as inside the liposomes.
  • composition of the present invention may be characterized by having a multi-lamellar vesicle (MLV) structure, but is not limited thereto.
  • MLV multi-lamellar vesicle
  • composition of the present invention may be characterized in that the surface charge value is a negative value, but is not limited thereto.
  • Another aspect of the present invention pertains to a pharmaceutical composition for preventing or treating a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection, including the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient.
  • prevention refers to any action that inhibits the above-mentioned disease or delays the progression thereof by administration of the pharmaceutical composition according to the present invention.
  • treatment refers to any action that ameliorates or advantageously changes symptoms of the above-mentioned disease by administration of the pharmaceutical composition according to the present invention.
  • the pharmaceutical composition of the present invention may be included in combination with at least one pharmaceutically or physiologically acceptable carrier, diluent, or excipient.
  • the pharmaceutical composition may include buffers such as neutral buffered saline, phosphate buffered saline, citrate buffer, etc.; carbohydrates such as glucose, mannose, sucrose, dextran, or mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g. aluminum hydroxide); and preservatives.
  • the pharmaceutical composition of the present invention may be administered orally or parenterally, for example through intravenous administration, subcutaneous administration, intradermal administration, intramuscular administration, intraperitoneal administration, intratumoral administration, intracerebral administration, intracranial administration, intrapulmonary administration, and intrarectal administration, but the present invention is not limited thereto.
  • the pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount.
  • a "pharmaceutically effective amount” is an amount sufficient to treat a disease at a reasonable benefit/risk ratio applicable to medical treatment, and the effective amount may be determined depending on the type of disease of a patient, severity of disease, drug activity, drug sensitivity, time of administration, route of administration, excretion rate, duration of treatment, type of concomitant drug, and other factors well known in the medical field.
  • the pharmaceutical composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with a conventional therapeutic agent, and may be administered once or multiple times. Taking into consideration all of the above factors, it is important to administer an amount that may obtain the maximum effect in the minimum amount without side effects, which may be easily determined by those skilled in the art.
  • the effective amount of the pharmaceutical composition of the present invention may vary depending on the patient's age, gender, condition, body weight, absorption rate of the active ingredient in the body, inactivation rate, and excretion rate, type of disease, and type of concomitant drug.
  • the pharmaceutical composition of the present invention includes the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient, a redundant description thereof is omitted to avoid excessive complexity in the present specification.
  • Still another aspect of the present invention pertains to a method of preventing or treating cancer, a tumor, an autoimmune disease, an inflammatory disease, a viral infection, or a bacterial infection, including administering the composition for delivering mRNA containing a modified nucleotide according to the present invention to a patient in need of prevention or treatment for cancer, a tumor, an autoimmune disease, an inflammatory disease, a viral infection, or a bacterial infection.
  • the amount of mRNA that is administered may be 1 to 5 pg, 5 to 10 pg, 10 to 15 pg, 15 to 20 pg, 10 to 25 pg, 20 to 25 pg, 20 to 50 pg, 30 to 50 pg, 40 to 50 pg, 40 to 60 pg, 60 to 80 pg, 60 to 100 pg, 50 to 100 pg, 80 to 120 pg, 40 to 120 pg, 40 to 150 pg, 50 to 150 ⁇ g, 50 to 200 ⁇ g, 80 to 200 ⁇ g, 100 to 200 ⁇ g, 120 to 250 pg, 150 to 250 pg, 180 to 280 pg, 200 to 300 pg, 50 to 300 pg, 80 to 300 ⁇ g, 100 to 300 ⁇ g, 40 to 300 ⁇ g, 50 to 350 ⁇ g, 100 to 350 pg, 200 to 350 pg, 300 to 350 pg, 320 to 400 pg, 40 to 380 pg, 40 to
  • Yet another aspect of the present invention pertains to a vaccine including the composition for delivering mRNA containing a modified nucleotide as an active ingredient.
  • the vaccine may be a vaccine against viruses capable of infecting humans and animals, such as influenza, coronavirus, shingles, human papillomavirus, Zika virus, herpes virus, AIDS virus, SFTS virus, measles virus, varicella virus, Ebola virus, MERS virus, hepatitis virus, avian influenza, rabies virus, and foot-and-mouth disease virus, but the present invention is not limited thereto.
  • viruses capable of infecting humans and animals such as influenza, coronavirus, shingles, human papillomavirus, Zika virus, herpes virus, AIDS virus, SFTS virus, measles virus, varicella virus, Ebola virus, MERS virus, hepatitis virus, avian influenza, rabies virus, and foot-and-mouth disease virus, but the present invention is not limited thereto.
  • the vaccine may be a multivalent vaccine
  • the virus included in the multivalent vaccine may include two or more viruses selected from the group consisting of viruses capable of infecting humans and animals, such as influenza, coronavirus, shingles, human papillomavirus, Zika virus, herpes virus, AIDS virus, SFTS virus, measles virus, varicella virus, Ebola virus, MERS virus, hepatitis virus, avian influenza, rabies virus, and foot-and-mouth disease virus, but the present invention is not limited thereto.
  • viruses capable of infecting humans and animals such as influenza, coronavirus, shingles, human papillomavirus, Zika virus, herpes virus, AIDS virus, SFTS virus, measles virus, varicella virus, Ebola virus, MERS virus, hepatitis virus, avian influenza, rabies virus, and foot-and-mouth disease virus, but the present invention is not limited thereto.
  • the vaccine includes mRNA containing at least one modified nucleotide having an ORF encoding at least one viral antigen polypeptide or an immunogenic fragment thereof.
  • the vaccine may include the composition for delivering mRNA containing a modified nucleotide at an appropriate concentration in consideration of the body weight, age, dietary stage, and/or immunity of an administration subject within a range of the purpose of preventing a disease caused by a peptide or protein that may act as an immunogen by the aforementioned mRNA.
  • the vaccine may further include at least one selected from the group consisting of a carrier, a diluent, an excipient, and an adjuvant.
  • a carrier is not particularly limited, but may include any and all solvents, dispersion media, coatings, stabilizers, preservatives, antibacterial and antifungal agents, isotonic agents, absorption delaying agents, and the like.
  • the vaccine may be administered through oral, parenteral, subcutaneous, intramuscular, intradermal, sublingual, transdermal, intrarectal, transmucosal, surface area via inhalation, or buccal routes, or combinations thereof.
  • the vaccine may be administered once or several times, and also intermittently, for example daily for days, weeks, or months, in the same amount or in different amounts, depending on the desired duration and effectiveness of vaccination or treatment.
  • An injectable solution thereof may be administered by injection in a desired amount or by spraying subcutaneously or intranasally, or alternatively by continuous infusion.
  • the vaccine may be provided in the form of a lyophilized formulation, but is not limited thereto.
  • the vaccine of the present invention includes the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient, a redundant description thereof is omitted to avoid excessive complexity in the present specification.
  • Still yet another aspect of the present invention pertains to a functional cosmetic composition including the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient.
  • the cosmetic composition according to the present invention may include components commonly used in cosmetic compositions, for example, typical adjuvants such as sequestering agents, active ingredients (e.g. sodium hyaluronate, tocopheryl acetate, etc.), preservatives, thickeners, and fragrances, and carriers.
  • typical adjuvants such as sequestering agents, active ingredients (e.g. sodium hyaluronate, tocopheryl acetate, etc.), preservatives, thickeners, and fragrances, and carriers.
  • the cosmetic composition according to the present invention may be prepared in the form of a typical formulation in the art, for example, an emulsion formulation or a solubilization formulation.
  • the emulsion formulation may be exemplified by nutrient lotion, cream, essence, etc.
  • the solubilization formulation may be exemplified by softening lotion.
  • the cosmetic composition of the present invention may be prepared in the form of an adjuvant that may be applied topically or systemically, which is commonly used in the field of dermatology, by containing a dermatologically acceptable medium or base in addition to cosmetics.
  • suitable cosmetic formulations may include solutions, gels, solid or anhydrous paste products, oil-in-water emulsions, suspensions, microemulsions, microcapsules, microgranules, ionic (liposomes) or nonionic follicular dispersions, creams, toners, lotions, powders, ointments, sprays, and conceal sticks.
  • the cosmetic composition of the present invention may be prepared in the form of foam or an aerosol composition further containing a compressed propellant.
  • the cosmetic composition of the present invention may further include a fatty material, an organic solvent, a solubilizing agent, a thickening agent, a gelling agent, a softening agent, an antioxidant, a suspending agent, a stabilizer, a foaming agent, a fragrance, a surfactant, water, an ionic or nonionic emulsifier, a filler, a sequestering agent, a chelating agent, a preservative, a vitamin, a blocking agent, a humectant, an essential oil, a dye, a pigment, a hydrophilic or lipophilic activating agent, a lipid vesicle, or an adjuvant commonly used in cosmetology or dermatology, such as any other ingredient commonly used in cosmetics.
  • the above ingredients may be introduced in amounts generally used in the field of dermatology.
  • Examples of products to which the cosmetic composition of the present invention may be added may include cosmetics such as astringent lotions, softening lotions, nutrient lotions, various creams, essences, packs, foundations, etc., as well as cosmetics such as cleansers, face washes, soaps, treatments, beauty essences, etc.
  • cosmetics such as astringent lotions, softening lotions, nutrient lotions, various creams, essences, packs, foundations, etc.
  • cosmetics such as cleansers, face washes, soaps, treatments, beauty essences, etc.
  • Specific formulations of the cosmetic composition of the present invention include skin lotion, skin softener, skin toner, astringent, lotion, milk lotion, moisture lotion, nutrient lotion, massage cream, nutrient cream, moisture cream, hand cream, essence, nutrient essence, pack, soap, shampoo, cleansing foam, cleansing lotion, cleansing cream, body lotion, body cleanser, emulsion, pressed powder, loose powder, patch, spray, and the like.
  • the cosmetic composition of the present invention includes the composition for delivering mRNA containing a modified nucleotide described above as an active ingredient, a redundant description thereof is omitted to avoid excessive complexity in the present specification.
  • a further aspect of the present invention pertains to the use of the composition for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid for preventing or treating a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection.
  • compositions for delivering mRNA containing a modified nucleotide including liposomes based on a cationic lipid for the manufacture of a medicament for preventing or treating a disease selected from the group consisting of cancer, a tumor, an autoimmune disease, a genetic disease, an inflammatory disease, a viral infection, and a bacterial infection.
  • Modified nucleotides used were N1-methylpseudouridine (m1 ⁇ , TriLink BioTechnologies, USA) and 5-methoxyuridine (5moU, TriLink BioTechnologies, USA), and target mRNA templates used were Firefly Luciferase (F. Luc) and SARS-CoV-2 omicron spike protein mRNA (EG-COVARo).
  • mRNA containing a modified nucleotide was synthesized by substituting any uridine with the corresponding modified nucleotide.
  • synthesized mRNA are as follows, and the ORFs of Luciferase and SARS-CoV-2 omicron spike protein mRNA (EG-COVARo) used and the amino acid sequences constructed thereby are shown in Table 1 below.
  • DOTAP CH2900014, Merck, Germany
  • DOPE L-R4-069, Corden Pharma, Germany
  • cholesterol W004591, Sigma-Aldrich, USA
  • a lipid mixture was prepared by mixing the liquid solutions at a predetermined weight ratio in a round bottom flask, and the lipid mixture containing DOTAP was volatilized at 60°C for 30 minutes in a rotary evaporator (B491_R200, Buchi, Switzerland) to blow off chloroform, and a lipid membrane film was formed on the wall of the flask.
  • a rotary evaporator B491_R200, Buchi, Switzerland
  • Respective mRNA-liposome complexes were prepared by mixing the liposomes prepared in Example 2 (LP-DOTAP/DOPE/cholesterol (40:40:20)) and 6 types of mRNA in 20 mM HEPES buffer containing 4% sucrose.
  • the liposome solution (LP-DOTAP/DOPE/cholesterol (40:40:20)) used was one obtained immediately after preparation or refrigerated for up to 2 weeks after preparation.
  • the mRNA solution was stored at -70°C or less, and was used after thawing on ice immediately before use.
  • 20 mM HEPES buffer containing 4% sucrose used was one prepared immediately before the experiment or refrigerated (2-8°C) after preparation the day before the experiment.
  • the N/P ratio of cationic liposomes and each mRNA was 0.6:1.0.
  • mRNA of each of 2) and 3) prepared in Example 1 was mixed with cationic liposomes in the same manner as in Example 3 to form mRNA-liposome complexes.
  • mice were anesthetized by intraperitoneal administration of 250 mg/kg of Avertin working solution, and 20 mL of 1x PBS was added to a luciferase substrate (VivoGlo TM Luciferin, P1043, Promega, USA) to obtain a 50 mg/mL substrate working solution, which was then diluted to 15 mg/mL before administration, and 200 ⁇ L thereof per mouse was intravenously injected through the tail vein (IV injection).
  • a luciferase substrate VivoGlo TM Luciferin, P1043, Promega, USA
  • mice Immediately after substrate administration, the mice were placed in an in-vivo imaging system (IVIS) (Ami HTX, Spectral Instrument, USA), images were taken by setting the exposure time to 60 seconds, and the luciferase expression level of the region of interest (ROI) was quantified.
  • IVIS in-vivo imaging system
  • the ROI value of the 5moU modified mRNA + liposome group was confirmed to be about 4.3 to 10.2 times higher than that of the m1 ⁇ modified mRNA + liposome group.
  • mRNA of each of 5) and 6) prepared in Example 1 was mixed with cationic liposomes in the same manner as in Example 3 to prepare mRNA-liposome complexes.
  • mice Two weeks after final immunization, the mice were sacrificed, serum was separated, and the end-point titer was determined by analyzing the SARS-CoV-2 omicron receptor-binding domain (RBD) protein-specific total IgG antibody titer (logic) by indirect ELISA.
  • RBD SARS-CoV-2 omicron receptor-binding domain
  • mice Two weeks after the last administration of the mRNA-liposome complex, mice were anesthetized by intraperitoneal administration of 250 mg/kg of Avertin working solution, and whole blood was collected through cardiac blood sampling. The whole blood thus collected was transferred to a microtube, allowed to stand at room temperature for 3 hours, and centrifuged at 4°C and 15,000 rpm for 10 minutes, after which the supernatant was transferred to a new microtube and serum was obtained and stored at -20°C or less until analysis.
  • RBD antigen SARS-CoV-2 spike protein, MBS335825, Mybiosource, USA
  • 1x PBS 1x PBS
  • dispensed at 100 ⁇ L/well into an immunoplate covered with a sealing film, and allowed to stand overnight in a refrigerator at 4°C.
  • the solution in each well was removed with an ELISA washer (Hydroflexelisa, Tecan, Switzerland), followed by washing three times using wash buffer (addition of 500 ⁇ L of Tween-20 to 1 L of 1x PBS obtained by diluting 20x PBS with purified water).
  • a reagent diluent (1% BSA in PBS, prepared by dissolving 1 g of BSA in 100 mL of PBS) was dispensed at 200 ⁇ L/well into the immunoplate, covered with a sealing film, and allowed to stand in a reactor at 37°C for 1 hour. The solution in each well was removed with an ELISA washer, followed by washing three times using wash buffer. A reagent diluent was dispensed at 100 ⁇ L/well into the immunoplate.
  • a negative control group buffer administration group
  • a vaccine administration group at 1:100 among the serum samples obtained above using a reagent diluent (1% BSA in PBS)
  • 100 ⁇ L thereof was dispensed in line 1 of B to G of the immunoplate, and the sample was mixed by pipetting several times in the well, after which the sample was subjected to 1/2 serial dilution up to line 12 on the ELISA plate in a manner of taking 100 ⁇ L from line 1 and adding the same to line 2.
  • hyperserum was diluted 1:200 using a reagent diluent, after which 100 ⁇ L thereof was dispensed in line 1 of H of the immunoplate, followed by 1/2 serial dilution in the same manner as above.
  • the immunoplate was covered with a sealing film, followed by reaction in a reactor at 37°C for 2 hours.
  • the solution in each well was removed with an ELISA washer, followed by washing three times using wash buffer.
  • a goat anti-mouse IgG antibody (Jackson Laboratory, USA) was diluted 1:5,000 using a reagent diluent, dispensed in an amount of 100 ⁇ L into the immunoplate, covered with a sealing film, and allowed to react in a reactor at 37°C for 1 hour.
  • the solution in each well was removed with an ELISA washer, followed by washing three times using wash buffer.
  • a TMB substrate working solution equilibrated to room temperature was dispensed at 100 ⁇ L/well into the immunoplate and allowed to react for 12 minutes in the dark at room temperature. The reaction was stopped by dispensing a 1 N H 2 SO 4 solution at 100 ⁇ L/well into the immunoplate, and absorbance was measured at 450 nm using an ELISA reader (Epoch, Biotek, USA).
  • Example 6 Identification of inflammatory response depending on type of modified nucleotide
  • Example 1 In order to confirm the inflammatory response of the mRNA-liposome complex depending on the type of modified nucleotide, mRNA of each of 4), 5), and 6) prepared in Example 1 was mixed with cationic liposomes in the same manner as in Example 3 to form mRNA-liposome complexes.
  • Mouse immune cell line J774A.1 cells (TIB-67, ATCC, USA) were seeded at 1 ⁇ 10 6 cells/well in a 24-well cell culture plate, followed by culture overnight at 37°C in a CO 2 incubator. When cell confluency was 70-80%, the cells were treated with each mRNA-liposome complex at 10 ⁇ g/well based on mRNA, followed by culture overnight at 37°C in a CO 2 incubator. In a positive control group, the cells were treated with LPS at 10 ug/well. Thereafter, the plate was centrifuged at 3,000 rpm and 4°C for 3 minutes, after which the supernatant was decanted and each cell culture fluid was collected.
  • the concentration of inflammatory cytokine marker secreted in each culture fluid was measured using ELISA, and the ELISA kit used was as follows.
  • the secretion of IL-1 ⁇ , IL-6, TNF- ⁇ , and IFN- ⁇ in the mRNA using unmodified nucleotide + liposome group was about 1.9 to 159.5 times higher than that in the 5moU modified mRNA + liposome group, and also, the secretion of IL-6, TNF- ⁇ , and IFN- ⁇ in the m1 ⁇ modified mRNA + liposome group was about 2.5 to 94.3 times higher than that in the 5moU modified mRNA + liposome group.
  • the inflammatory response is low compared to when using an unmodified nucleotide due to the influence of noninflammatory mRNA.
  • the 5moU modified mRNA + liposome group showed a lower level of inflammatory cytokine secretion than the m1 ⁇ modified mRNA + liposome group, confirming that the inflammatory response was greatly reduced in the combination of 5moU modified mRNA and liposomes.
  • a composition for delivering mRNA containing a modified nucleotide has superior storage stability and exhibits high intracellular delivery and expression efficiencies in vivo, thereby increasing the stability and efficiency of mRNA vaccines for cancer treatment or mRNA vaccines for preventing viral infections and other mRNA vaccines or therapeutics.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dispersion Chemistry (AREA)
  • Molecular Biology (AREA)
  • Dermatology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
EP23814344.0A 2022-09-07 2023-09-06 Zusammensetzung zur in-vivo-freisetzung von mrna mit modifiziertem nukleotid Pending EP4356933A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
KR20220113631 2022-09-07
KR1020230102612A KR20240035322A (ko) 2022-09-07 2023-08-07 변형핵산 함유 mRNA의 체내 전달용 조성물
PCT/KR2023/013302 WO2024054020A1 (ko) 2022-09-07 2023-09-06 변형핵산 함유 mrna의 체내 전달용 조성물

Publications (1)

Publication Number Publication Date
EP4356933A1 true EP4356933A1 (de) 2024-04-24

Family

ID=90191529

Family Applications (1)

Application Number Title Priority Date Filing Date
EP23814344.0A Pending EP4356933A1 (de) 2022-09-07 2023-09-06 Zusammensetzung zur in-vivo-freisetzung von mrna mit modifiziertem nukleotid

Country Status (3)

Country Link
EP (1) EP4356933A1 (de)
AU (1) AU2023274159A1 (de)
WO (1) WO2024054020A1 (de)

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI3611266T1 (sl) 2005-08-23 2023-02-28 The Trustees Of The University Of Pennsylvania Modificirani nukleozidi, ki vsebujejo RNA, in postopki za njihovo uporabo
JP2013512690A (ja) 2009-12-07 2013-04-18 ザ トラスティース オブ ザ ユニバーシティ オブ ペンシルベニア 細胞を再プログラム化するための精製された修飾rnaを含むrna調製物
BR112013031553A2 (pt) * 2011-06-08 2020-11-10 Shire Human Genetic Therapies, Inc. composições, mrna que codifica para uma hgla e seu uso, uso de pelo menos uma molécula de mrna e um veículo de transferência e uso de um mrna que codifica para proteína exógena
AU2013243946A1 (en) * 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of membrane proteins
KR101509456B1 (ko) 2013-10-04 2015-04-14 아이진 주식회사 리포폴리사카라이드 유사체 및 이를 포함하는 면역보조 조성물
KR102096796B1 (ko) * 2013-10-22 2020-05-27 샤이어 휴먼 지네틱 테라피즈 인크. 메신저 rna의 전달을 위한 지질 제형
KR102042993B1 (ko) 2016-10-31 2019-11-11 아이진 주식회사 면역반응 조절물질 및 이를 포함하는 면역보조제 조성물
WO2018160592A1 (en) * 2017-02-28 2018-09-07 Arcturus Therapeutics, Inc. Translatable molecules and synthesis thereof
KR102014601B1 (ko) 2017-10-19 2019-08-26 인하대학교 산학협력단 3차원 유로구조 미세 유체 혼합기
KR20220117133A (ko) * 2021-02-15 2022-08-23 주식회사 바이오파마 양이온성 분자 수송체 및 SARS-CoV-2 mRNA의 이온 복합체를 포함하는 코로나바이러스감염증-19 예방 백신 조성물

Also Published As

Publication number Publication date
AU2023274159A1 (en) 2024-03-21
WO2024054020A1 (ko) 2024-03-14

Similar Documents

Publication Publication Date Title
EP3917503B1 (de) Verfahren zur herstellung von lipidnanopartikeln
US20240009131A1 (en) Methods of making lipid nanoparticles
US20230364024A1 (en) Stabilized formulations of lipid nanoparticles
US20210378980A1 (en) Preparation of lipid nanoparticles and methods of administration thereof
JP2022095702A (ja) 脂質ナノ粒子の安定化製剤
TW202139976A (zh) 製備脂質奈米顆粒之方法
US20230053437A1 (en) Lipid compounds and lipid nanoparticle compositions
WO2022152141A2 (en) Polymer conjugated lipid compounds and lipid nanoparticle compositions
EP4306132A1 (de) Zusammensetzung zur in-vivo-freisetzung von rna und herstellungsverfahren dafür
EP4356933A1 (de) Zusammensetzung zur in-vivo-freisetzung von mrna mit modifiziertem nukleotid
KR20240035322A (ko) 변형핵산 함유 mRNA의 체내 전달용 조성물
TW202410883A (zh) 用於體內遞送包含經修飾的核苷酸的mRNA的組合物
WO2024083172A1 (zh) 脂质化合物和脂质纳米颗粒组合物
WO2024022263A1 (zh) 脂质化合物和脂质纳米颗粒组合物
WO2024109798A1 (zh) 脂质化合物和脂质纳米颗粒组合物
WO2024037577A1 (en) Composition of lipid nanoparticles
WO2023116804A1 (zh) 脂质化合物和脂质纳米颗粒组合物
CN117159492A (zh) 一种含有唾液酸脂质衍生物的脂质纳米颗粒及其应用
CN117069785A (zh) 脂质化合物和脂质纳米颗粒组合物

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20231208

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC ME MK MT NL NO PL PT RO RS SE SI SK SM TR